1
|
Tiselius E, Sundberg E, Andersson H, Höbinger A, Jahnmatz P, Harila A, Palle J, Nilsson A, Saghafian-Hedengren S. Bone Marrow-Suppressive Treatment in Children Is Associated with Diminished IFN-γ Response from T Cells upon Polyclonal and Varicella Zoster Virus Peptide Stimulation. Int J Mol Sci 2024; 25:6960. [PMID: 39000070 PMCID: PMC11241059 DOI: 10.3390/ijms25136960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Severe haematological diseases and lymphoid malignancies require bone marrow (BM)-suppressive treatments. Knowledge regarding the impact of BM-suppressive treatments on children's memory T cells is very limited. Memory T cells play a crucial role in defending against herpesviruses, which is particularly relevant in paediatric cancer care. We studied 53 children in total; 34 with cancer and 2 with severe haematological disorders, with some receiving BM-suppressive treatment with or without allogeneic-haematopoietic stem cell transplantation (allo-HSCT), alongside 17 healthy controls. We focused on peripheral blood proportions of memory T-cell subsets using flow cytometry and analysed cytokine-secreting T cells with a four-parameter FluoroSpot assay in response to T-cell mitogen and varicella zoster virus (VZV) peptides. Patients on BM-suppressive treatment showed increased clusters of differentiation (CD)4+ and CD8+ effector memory (TEM)/terminally differentiated effector (TEFF) T cells compared to the healthy controls. They also exhibited, amongst other things, when compared to the healthy controls, a reduced total number of cytokine-secreting cells, by means of interferon (IFN)-γ, interleukin (IL)-17A, IL-10, and IL-22, following mitogen activation. A diminished IFN-γ response among the children with BM-suppressive treatment was observed upon VZV-peptide stimulation, compared to the healthy children. Collectively, the findings herein indicate that the children who are undergoing or have finished BM-suppressive treatment display qualitative differences in their T-cell memory compartment, potentially increasing their susceptibility to severe viral infections and impacting their immunotherapy, which relies on the functional ability of autologous T cells.
Collapse
Affiliation(s)
- Eva Tiselius
- Department of Women’s and Children’s Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.T.); (S.S.-H.)
| | - Emil Sundberg
- Department of Women’s and Children’s Health, Uppsala University, 751 05 Uppsala, Sweden; (E.S.); (A.H.); (J.P.)
| | - Hanna Andersson
- Department of Women’s and Children’s Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.T.); (S.S.-H.)
| | - Anna Höbinger
- Department of Women’s and Children’s Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.T.); (S.S.-H.)
| | | | - Arja Harila
- Department of Women’s and Children’s Health, Uppsala University, 751 05 Uppsala, Sweden; (E.S.); (A.H.); (J.P.)
- Department of Children’s Oncology and Hematology, Uppsala University Hospital, 751 85 Uppsala, Sweden
| | - Josefine Palle
- Department of Women’s and Children’s Health, Uppsala University, 751 05 Uppsala, Sweden; (E.S.); (A.H.); (J.P.)
- Department of Children’s Oncology and Hematology, Uppsala University Hospital, 751 85 Uppsala, Sweden
| | - Anna Nilsson
- Department of Women’s and Children’s Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.T.); (S.S.-H.)
| | - Shanie Saghafian-Hedengren
- Department of Women’s and Children’s Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, 171 77 Stockholm, Sweden; (E.T.); (S.S.-H.)
| |
Collapse
|
2
|
Antikainen E, Grönroos M, Huurre A, Korhonen L, Peltola V, Lähteenmäki P, Schuez‐Havupalo L. Treatment intensity affects immune reconstitution even after childhood cancer not treated with hematopoietic stem cell transplantation. Cancer Rep (Hoboken) 2024; 7:e2069. [PMID: 38767518 PMCID: PMC11104287 DOI: 10.1002/cnr2.2069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/09/2024] [Accepted: 03/23/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Only a few previous studies examine immune system recovery after completed cancer treatment. AIMS The aim of this study was to analyze immune reconstitution after childhood cancer therapy in a non-hematopoietic stem cell transplantation setting. METHODS AND RESULTS We analyzed children (N = 79) who received chemotherapy with/without irradiation for cancer diagnosed between 2014 and 2019 at Turku University Hospital, Finland. We retrospectively collected data on baseline parameters and post-treatment immunological recovery, namely neutrophil and lymphocyte counts, IgG levels, CD19, CD4 and natural killer cell counts. Immunological parameters were followed until their normalization. Treatment intensity was stratified according to the Intensity of Treatment Rating Scale (ITR-3). We analyzed the effects of treatment intensity on normalization of immunological parameters across the entire treatment range. Treatment intensity had a major effect on immune system recovery after completion of treatment. Most patients had normal immunological parameters 1-4 months post-treatment both in high- and low-intensity treatment groups, but patients classified in the high-intensity group had low parameters more often than patients in the low-intensity group. CONCLUSION Our data suggest a fast recovery of studied immunological parameters after the majority of current pediatric oncologic treatments. Treatment for high-risk acute lymphoblastic leukemia, acute myeloid leukemia, medulloblastoma, and mature B-cell lymphoma was associated with prolonged recovery times for a substantial proportion of cases. High treatment intensity was associated with prolonged immunological recovery.
Collapse
Affiliation(s)
| | - Marika Grönroos
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Anu Huurre
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Laura Korhonen
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Ville Peltola
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Päivi Lähteenmäki
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Linnea Schuez‐Havupalo
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| |
Collapse
|
3
|
Óskarsson Ý, Thors V, Vias RD, Lúðvíksson BR, Brynjólfsson SF, Gianchecchi E, Razzano I, Montomoli E, Gísli Jónsson Ó, Haraldsson Á. Adequate immune responses to vaccines after chemotherapy for leukaemia diagnosed in childhood. Acta Paediatr 2024; 113:606-614. [PMID: 38140806 DOI: 10.1111/apa.17070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/17/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
AIM The survival rate after treatment for childhood leukaemia has greatly improved, but could result in protracted immune deficiency. This study examined the immune status of children after chemotherapy and evaluated their responses to immunisation. METHODS Subjects who had completed their treatment for acute lymphoblastic leukaemia at The Children's Hospital Reykjavík, Iceland, during 2011-2020 had blood drawn and were then immunised for influenza in October 2021. Blood was drawn again 4 weeks later and their humoral and cellular responses were measured with a haemagglutination inhibition assay and lymphocyte stimulation test. Antibodies to other immunisations were also evaluated. RESULTS We studied 18 patients (10 male) who had completed their treatment at 3.7-20.3 years of age (mean 9.1), 11-84 months (mean 36.9) before enrolment. Conventional immunological evaluation did not reveal notable abnormalities. The responses to several childhood vaccinations, including the pneumococcal conjugate vaccination, were adequate in most patients. Humoral responses to the influenza vaccine confirmed adequate reactions in all but one patient. Considerable variations were observed in the lymphocyte stimulations tests. CONCLUSION Most patients reacted adequately to immunisation, especially against annual influenza and Streptococcus pneumoniae, reiterating the usefulness of vaccinations. The most appropriate timing for vaccination after treatment still needs to be determined.
Collapse
Affiliation(s)
- Ýmir Óskarsson
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Valtýr Thors
- The Children's Hospital Iceland, Landspítali University Hospital, Reykjavík, Iceland
- Department of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Rafael Daníel Vias
- Department of Mathematics, Faculty of Physical Sciences, University of Iceland, Reykjavík, Iceland
| | - Björn Rúnar Lúðvíksson
- Department of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
- Department of Immunology, Landspítali University Hospital, Reykjavík, Iceland
| | - Siggeir Fannar Brynjólfsson
- Department of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
- Department of Immunology, Landspítali University Hospital, Reykjavík, Iceland
| | | | | | - Emanuele Montomoli
- VisMederi Srl, Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ólafur Gísli Jónsson
- The Children's Hospital Iceland, Landspítali University Hospital, Reykjavík, Iceland
- Department of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Ásgeir Haraldsson
- The Children's Hospital Iceland, Landspítali University Hospital, Reykjavík, Iceland
- Department of Medicine, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
4
|
Elitzur S, Vora A, Burkhardt B, Inaba H, Attarbaschi A, Baruchel A, Escherich G, Gibson B, Liu HC, Loh M, Moorman AV, Möricke A, Pieters R, Uyttebroeck A, Baird S, Bartram J, Barzilai-Birenboim S, Batra S, Ben-Harosh M, Bertrand Y, Buitenkamp T, Caldwell K, Drut R, Geerlinks AV, Gilad G, Grainger J, Haouy S, Heaney N, Huang M, Ingham D, Krenova Z, Kuhlen M, Lehrnbecher T, Manabe A, Niggli F, Paris C, Revel-Vilk S, Rohrlich P, Sinno MG, Szczepanski T, Tamesberger M, Warrier R, Wolfl M, Nirel R, Izraeli S, Borkhardt A, Schmiegelow K. EBV-driven lymphoid neoplasms associated with pediatric ALL maintenance therapy. Blood 2023; 141:743-755. [PMID: 36332176 DOI: 10.1182/blood.2022016975] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/19/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
The development of a second malignancy after the diagnosis of childhood acute lymphoblastic leukemia (ALL) is a rare event. Certain second malignancies have been linked with specific elements of leukemia therapy, yet the etiology of most second neoplasms remains obscure and their optimal management strategies are unclear. This is a first comprehensive report of non-Hodgkin lymphomas (NHLs) following pediatric ALL therapy, excluding stem-cell transplantation. We analyzed data of patients who developed NHL following ALL diagnosis and were enrolled in 12 collaborative pediatric ALL trials between 1980-2018. Eighty-five patients developed NHL, with mature B-cell lymphoproliferations as the dominant subtype (56 of 85 cases). Forty-six of these 56 cases (82%) occurred during or within 6 months of maintenance therapy. The majority exhibited histopathological characteristics associated with immunodeficiency (65%), predominantly evidence of Epstein-Barr virus-driven lymphoproliferation. We investigated 66 cases of post-ALL immunodeficiency-associated lymphoid neoplasms, 52 from our study and 14 additional cases from a literature search. With a median follow-up of 4.9 years, the 5-year overall survival for the 66 patients with immunodeficiency-associated lymphoid neoplasms was 67.4% (95% confidence interval [CI], 56-81). Five-year cumulative risks of lymphoid neoplasm- and leukemia-related mortality were 20% (95% CI, 10.2-30) and 12.4% (95% CI, 2.7-22), respectively. Concurrent hemophagocytic lymphohistiocytosis was associated with increased mortality (hazard ratio, 7.32; 95% CI, 1.62-32.98; P = .01). A large proportion of post-ALL lymphoid neoplasms are associated with an immunodeficient state, likely precipitated by ALL maintenance therapy. Awareness of this underrecognized entity and pertinent diagnostic tests are crucial for early diagnosis and optimal therapy.
Collapse
Affiliation(s)
- Sarah Elitzur
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ajay Vora
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Birgit Burkhardt
- Pediatric Hematology and Oncology, University Hospital Münster, Münster, Germany
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Andre Baruchel
- Department of Pediatric Hematology, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncoogy, University Medical Centre, Hamburg-Eppendorf, Hamburg, Germany
| | - Brenda Gibson
- Department of Paediatric Haematology, Royal Hospital for Children, Glasgow, United Kingdom
| | - Hsi-Che Liu
- Division of Pediatric Hematology/Oncology, Mackay Children's Hospital and Mackay Medical College, Taipei, Taiwan
| | - Mignon Loh
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant and Cellular Therapy, Seattle Children's Hospital and the Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA
| | - Anthony V Moorman
- Leukaemia Research Cytogenetics Group, Wolfson Childhood Cancer Centre, Clinical and Translational Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anja Möricke
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Anne Uyttebroeck
- Department of Paediatric Haematology and Oncology, University Hospital Leuven, Leuven, Leuven, Belgium
| | - Susan Baird
- Department of Haematology, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Jack Bartram
- Department of Paediatric Haematology, Great Ormond Street Hospital, London, United Kingdom
| | - Shlomit Barzilai-Birenboim
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Sandeep Batra
- Pediatric Hematology/Oncology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN
| | - Miriam Ben-Harosh
- Department of Pediatric Hemato-Oncology, Soroka Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yves Bertrand
- Institut d'Hematologie et d'Oncologie Pediatrique, Hospices Civils de Lyon, Lyon, France
| | - Trudy Buitenkamp
- Amsterdam Academic Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Kenneth Caldwell
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, FL
| | - Ricardo Drut
- Department of Pathology, School of Medicine, La Plata National University, La Plata, Argentina
| | | | - Gil Gilad
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - John Grainger
- Faculty of Medical & Human Sciences, University of Manchester and Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie Haouy
- Department of Pediatric Oncology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Nicholas Heaney
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Mary Huang
- Department of Pediatric Hematology Oncology, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA
| | - Danielle Ingham
- Paediatric Oncology, Leeds Children's Hospital, Leeds, United Kingdom
| | - Zdenka Krenova
- Department of Pediatric Oncology and Department of Pediatrics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michaela Kuhlen
- Pediatrics and Adolescent Medicine, University of Augsburg, Augsburg, Germany
| | - Thomas Lehrnbecher
- Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University, Graduate School of Medicine, Sapporo, Japan
| | - Felix Niggli
- Department of Pediatric Oncology, University Children's Hospital, Zurich, Switzerland
| | - Claudia Paris
- Department of Pediatric Oncology and Hematology, Hospital Luis Calvo Mackenna, Santiago, Chile
| | - Shoshana Revel-Vilk
- Shaare Zedek Medical Centre and The Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | | | - Mohamad G Sinno
- Phoenix Children's Hospital, Center for Cancer and Blood Disorders, Phoenix, AZ
| | - Tomasz Szczepanski
- Department of Pediatric Hematology and Oncology, Zabrze and Medical University of Silesia, Katowice, Poland
| | - Melanie Tamesberger
- Department of Pediatrics and Adolescent Medicine, Kepler University Clinic, Linz, Austria
| | | | - Matthias Wolfl
- Pediatric Oncology, Hematology and Stem Cell Transplantation Program, University Children's Hospital Würzburg, Würzburg, Germany
| | - Ronit Nirel
- Department of Statistics and Data Science, Hebrew University, Jerusalem, Israel
| | - Shai Izraeli
- Department of Pediatric Hematology and Oncology, Schneider Children's Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, The University Hospital, Rigshospitalet, and Institute of Clinical Medicine, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Arakawa Y, Hasegawa D, Miyamura T, Ohshima J, Kimura S, Imamura T, Koga Y, Yamamoto S, Ogawa A, Shinoda K, Eguchi M, Hosoi H, Imai K, Koh K, Tomizawa D. Postchemotherapy immune status in infants with acute lymphoblastic leukemia: A report from the JPLSG MLL-10 trial. Pediatr Blood Cancer 2022; 69:e29772. [PMID: 35796397 DOI: 10.1002/pbc.29772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 11/08/2022]
Abstract
The MLL-10 trial (UMIN000004801) modified a Children's Oncology Group (COG) AALL0631 therapy for infants with KMT2A-rearranged acute lymphoblastic leukemia (ALL). In 2016, one registered case developed secondary immunodeficiency during maintenance therapy and eventually died due to cytomegalovirus infection. Around the same time, fatal secondary immunodeficiencies were reported in five infants with ALL in North America who had received COG-based chemotherapy between 1996 and 2015. Given these cases, we decided to conduct a retrospective study on the postchemotherapy immune status of infants with ALL. A questionnaire collected data on posttreatment immune function, frequency of infections, and supportive care for the 34 infants in the MLL-10 trial. Patients receiving allogeneic hematopoietic stem cell transplantation in first remission were excluded. Responses to the survey were obtained in 28 cases (85%). Most patients were immunocompetent after the completion of chemotherapy (median follow-up duration from the day of chemotherapy completion was 431 days), except for the aforementioned case. There were seven patients with nonsevere viral infection, all of whom recovered. In conclusion, severe chemotherapy-induced immunodeficiency in infants with ALL appears to be rare, but prospective data collection of immune function is necessary to clarify this finding.
Collapse
Affiliation(s)
- Yuki Arakawa
- Department of Haematology and Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Takako Miyamura
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Junjiro Ohshima
- Department of Pediatrics, Matsumoto Pediatrics, Sapporo, Japan.,Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Kimura
- Department of Pediatrics, St. Luke's International Hospital, Tokyo, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuhki Koga
- Department of Pediatrics, Kyushu University Hospital, Fukuoka, Japan
| | | | - Atsushi Ogawa
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | | | - Mariko Eguchi
- Department of Pediatrics, Ehime University Graduate School of Medicine, Tōon, Ehime, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohsuke Imai
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuyoshi Koh
- Department of Haematology and Oncology, Saitama Children's Medical Center, Saitama, Japan
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
6
|
Titov A, Kaminskiy Y, Ganeeva I, Zmievskaya E, Valiullina A, Rakhmatullina A, Petukhov A, Miftakhova R, Rizvanov A, Bulatov E. Knowns and Unknowns about CAR-T Cell Dysfunction. Cancers (Basel) 2022; 14:1078. [PMID: 35205827 PMCID: PMC8870103 DOI: 10.3390/cancers14041078] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/29/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR) T cells is a promising option for cancer treatment. However, T cells and CAR-T cells frequently become dysfunctional in cancer, where numerous evasion mechanisms impair antitumor immunity. Cancer frequently exploits intrinsic T cell dysfunction mechanisms that evolved for the purpose of defending against autoimmunity. T cell exhaustion is the most studied type of T cell dysfunction. It is characterized by impaired proliferation and cytokine secretion and is often misdefined solely by the expression of the inhibitory receptors. Another type of dysfunction is T cell senescence, which occurs when T cells permanently arrest their cell cycle and proliferation while retaining cytotoxic capability. The first section of this review provides a broad overview of T cell dysfunctional states, including exhaustion and senescence; the second section is focused on the impact of T cell dysfunction on the CAR-T therapeutic potential. Finally, we discuss the recent efforts to mitigate CAR-T cell exhaustion, with an emphasis on epigenetic and transcriptional modulation.
Collapse
Affiliation(s)
- Aleksei Titov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, 125167 Moscow, Russia
| | - Yaroslav Kaminskiy
- Laboratory of Transplantation Immunology, National Research Centre for Hematology, 125167 Moscow, Russia
| | - Irina Ganeeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Ekaterina Zmievskaya
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aygul Valiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aygul Rakhmatullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Alexey Petukhov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Institute of Hematology, Almazov National Medical Research Center, 197341 Saint Petersburg, Russia
| | - Regina Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
7
|
Ghassemi S, Durgin JS, Nunez-Cruz S, Patel J, Leferovich J, Pinzone M, Shen F, Cummins KD, Plesa G, Cantu VA, Reddy S, Bushman FD, Gill SI, O'Doherty U, O'Connor RS, Milone MC. Rapid manufacturing of non-activated potent CAR T cells. Nat Biomed Eng 2022; 6:118-128. [PMID: 35190680 PMCID: PMC8860360 DOI: 10.1038/s41551-021-00842-6] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022]
Abstract
Chimaeric antigen receptor (CAR) T cells can generate durable clinical responses in B-cell haematologic malignancies. The manufacturing of these T cells typically involves their activation, followed by viral transduction and expansion ex vivo for at least 6 days. However, the activation and expansion of CAR T cells leads to their progressive differentiation and the associated loss of anti-leukaemic activity. Here we show that functional CAR T cells can be generated within 24 hours from T cells derived from peripheral blood without the need for T-cell activation or ex vivo expansion, and that the efficiency of viral transduction in this process is substantially influenced by the formulation of the medium and the surface area-to-volume ratio of the culture vessel. In mouse xenograft models of human leukaemias, the rapidly generated non-activated CAR T cells exhibited higher anti-leukaemic in vivo activity per cell than the corresponding activated CAR T cells produced using the standard protocol. The rapid manufacturing of CAR T cells may reduce production costs and broaden their applicability.
Collapse
Affiliation(s)
- Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Joseph S Durgin
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jai Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Leferovich
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marilia Pinzone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Shen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine D Cummins
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vito Adrian Cantu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantan Reddy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saar I Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Una O'Doherty
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Chu X, Qian M, Yang J, Wu D, Gao J, Cao L, Fang F, Pan J, Zhang H, Hu S. Effect of GATA3 rs3824662 gene polymorphism in Han Chinese children with pre-B-cell acute lymphoblastic leukemia with 10 years follow-up. Front Pediatr 2022; 10:1044866. [PMID: 36714653 PMCID: PMC9875006 DOI: 10.3389/fped.2022.1044866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
PURPOSE To evaluate the influence of GATA3 rs3824662 on pre-B-cell acute lymphoblastic leukemia (pre-B-cell ALL) susceptibility and long-term prognosis in Han Chinese children with pre-B-cell ALL treated with the CCLG-2008 protocol at the Children's Hospital of Soochow University. METHODS A total of 256 patients with childhood pre-B-cell ALL under the CCLG-2008 protocol were enrolled in this study, and 174 healthy children were used as case controls. GATA3 rs3824662 genotyping was performed using a polymerase chain reaction, followed by Sanger sequencing. The association of genotype with clinical characteristics, treatment response, adverse events, and outcomes were analyzed. RESULTS The A allele frequency of GATA3 rs3824662 in patients with pre-B cell ALL was significantly higher than that in healthy children (OR = 1.41, 95% CI = 1.042-1.908; P = 0.026). Among patients with pre-B-cell ALL, the GATA3 rs3824662 AA genotype was associated with poor prednisolone response and high blast cell burden on day 15 of the induction therapy (P = 0.011 and 0.007, respectively). Patients with the rs3824662 AA variant suffered more episodes of sepsis than those with the CC or CA variants (P = 0.021). The GATA3 rs3824662 AA genotype was significantly associated with sepsis [hazard ratio (HR) = 3.375; P = 0.01]. No significant differences were found in the cumulative incidence of relapse, overall survival, and event-free survival among all genotypes. CONCLUSION GATA3 rs3824662 was associated with susceptibility in Han Chinese children with pre-B-cell ALL and could be a possible risk factor for poor early treatment response and treatment-related sepsis.
Collapse
Affiliation(s)
- Xinran Chu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Maoxiang Qian
- Department of Hematology and Oncology, Institute of Biomedical Sciences, Children's Hospital of Fudan University, Shanghai, China
| | - Jin Yang
- Department of Pediatrics, Subei People's Hospital of Jiangsu Province, Yangzhou, China
| | - Dong Wu
- Department of Pediatrics, Yiyuan People's Hospital, Zibo, China
| | - Jing Gao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| | - Lu Cao
- Department of Emergency, Children's Hospital of Soochow University, Suzhou, China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Hui Zhang
- Department of Hematology and Oncology, Fujian Branch of Shanghai Children's Medical Center, Fujian Children's Hospital, Fuzhou, China.,Department of Hematology and Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
9
|
Lingering effects of chemotherapy on mature T cells impair proliferation. Blood Adv 2021; 4:4653-4664. [PMID: 33002133 DOI: 10.1182/bloodadvances.2020001797] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Engineered T-cell therapies have demonstrated impressive clinical responses in patients with hematologic malignancies. Despite this efficacy, many patients have a transient persistence of T cells, which can be correlated with transient clinical response. Translational data on T cells from pediatric cancer patients shows a progressive decline in chimeric antigen receptor (CAR) suitability with cumulative chemotherapy regardless of regimen. We investigated the effects of chemotherapy on surviving T cells in vitro, describing residual deficits unique to each agent including mitochondrial damage and metabolic alterations. In the case of cyclophosphamide but not doxorubicin or cytarabine, these effects could be reversed with N-acetylcysteine. Specifically, we observed that surviving T cells could be stimulated, expanded, and transduced with CARs with preserved short-term cytolytic function but at far lower numbers and with residual metabolic deficits. These data have implications for understanding the effects of chemotherapy on mature T cells later collected for adoptive cell therapy, as chemotherapy-exposed T cells may have lingering dysfunction that affects ex vivo adoptive cell therapy manufacturing techniques and, ultimately, clinical efficacy.
Collapse
|
10
|
Saghafian-Hedengren S, Sverremark-Ekström E, Nilsson A. T Cell Subsets During Early Life and Their Implication in the Treatment of Childhood Acute Lymphoblastic Leukemia. Front Immunol 2021; 12:582539. [PMID: 33763058 PMCID: PMC7982872 DOI: 10.3389/fimmu.2021.582539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
The immune system plays a major role in recognizing and eliminating malignant cells, and this has been exploited in the development of immunotherapies aimed at either activating or reactivating the anti-tumor activity of a patient's immune system. A wide range of therapeutic approaches involving T lymphocytes, such as programmed cell death protein ligand-1 (PDL-1) inhibitors, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) blockers, and CD19-targeted T-cell therapy through chimeric antigen receptor (CAR)-T cells or CD19/CD3 bi-specific T-cell engagers, have been introduced to the field of oncology, leading to significant improvements in overall survival of adult cancer patients. During the past few years, the availability and approval of T-cell based immunotherapies have become a reality also for the treatment of childhood cancers. However, the distribution, ratio of regulatory to effector cells and the quality of T-cell responses early in life are distinct from those during adolescence and adulthood, raising the possibility that these differences impact the efficacy of immunotherapy. Herein we provide a brief overview of the properties of conventional T cell subsets during early life. Focusing on the most common cancer type during childhood, acute lymphoblastic leukemia (ALL), we describe how current conventional therapies used against ALL influence the T-cell compartment of small children. We describe early life T-cell responses in relation to immunotherapies engaging T-cell anticancer reactivity and present our opinion that it is not only immaturity of the adaptive immune system, but also the impact of an immunosuppressive environment that may prove disadvantageous in the setting of immunotherapies targeting pediatric cancer cells.
Collapse
Affiliation(s)
- Shanie Saghafian-Hedengren
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna Nilsson
- Division of Paediatric Oncology and Paediatric Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Jiang J, Ahuja S. Addressing Patient to Patient Variability for Autologous CAR T Therapies. J Pharm Sci 2021; 110:1871-1876. [PMID: 33340532 DOI: 10.1016/j.xphs.2020.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy clinical trials have had unprecedented success in the endeavors to cure cancer patients, particularly those having hematological cancers. As researchers learn more about the ways to make CAR T cells more effective to kill tumor cells, equally important will be understanding the differences between T cells from healthy donors and cancer patients and how these differences could affect ex vivo expansion of T cells during CAR T production. This undoubtedly could be a crucial factor in treating solid tumors, where CAR T cells are needed in significantly higher numbers. As the evidence for significant differences between the patients and healthy donors is compelling, an adaptable and robust production process should be designed to allow manufacture of the required CAR T cells for all cancer patients. Improving the fundamental understanding of the cellular metabolism and accompanying epigenetic and phenotypic changes during in vivo and ex vivo expansion of T cells will be just as important. Such discoveries will provide an invaluable tool box from which actionable knowledge could be drawn for designing an adaptable CAR T production process that is able to absorb the patient-to-patient variation.
Collapse
Affiliation(s)
- Jinlin Jiang
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD
| | - Sanjeev Ahuja
- Cell Culture and Fermentation Sciences, BioPharmaceutical Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD.
| |
Collapse
|
12
|
Bailur JK, McCachren SS, Pendleton K, Vasquez JC, Lim HS, Duffy A, Doxie DB, Kaushal A, Foster C, DeRyckere D, Castellino S, Kemp ML, Qiu P, Dhodapkar MV, Dhodapkar KM. Risk-associated alterations in marrow T cells in pediatric leukemia. JCI Insight 2020; 5:140179. [PMID: 32692727 PMCID: PMC7455136 DOI: 10.1172/jci.insight.140179] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
Abstract
Current management of childhood leukemia is tailored based on disease risk determined by clinical features at presentation. Whether properties of the host immune response impact disease risk and outcome is not known. Here, we combine mass cytometry, single cell genomics, and functional studies to characterize the BM immune environment in children with B cell acute lymphoblastic leukemia and acute myelogenous leukemia at presentation. T cells in leukemia marrow demonstrate evidence of chronic immune activation and exhaustion/dysfunction, with attrition of naive T cells and TCF1+ stem-like memory T cells and accumulation of terminally differentiated effector T cells. Marrow-infiltrating NK cells also exhibit evidence of dysfunction, particularly in myeloid leukemia. Properties of immune cells identified distinct immune phenotype–based clusters correlating with disease risk in acute lymphoblastic leukemia. High-risk immune signatures were associated with expression of stem-like genes on tumor cells. These data provide a comprehensive assessment of the immune landscape of childhood leukemias and identify targets potentially amenable to therapeutic intervention. These studies also suggest that properties of the host response with depletion of naive T cells and accumulation of terminal-effector T cells may contribute to the biologic basis of disease risk. Properties of immune microenvironment identified here may also impact optimal application of immune therapies, including T cell–redirection approaches in childhood leukemia. In high risk pediatric leukemia, naive and stem-like memory T cells are depleted and terminally differentiated effector T cells accumulate.
Collapse
Affiliation(s)
- Jithendra Kini Bailur
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Samuel S McCachren
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA.,The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Katherine Pendleton
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Juan C Vasquez
- Yale University School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Hong Seo Lim
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Alyssa Duffy
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Deon B Doxie
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Akhilesh Kaushal
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Connor Foster
- Yale University School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Sharon Castellino
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | - Melissa L Kemp
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Peng Qiu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Madhav V Dhodapkar
- Department of Hematology/Oncology, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA.,Winship Cancer Institute, Atlanta, Georgia, USA
| | - Kavita M Dhodapkar
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA.,Winship Cancer Institute, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Wu J, Wang X, Lin L, Li X, Liu S, Zhang W, Luo L, Wan Z, Fang M, Zhao Y, Wang X, Mai H, Yuan X, Wen F, Li C, Liu X. Developing an Unbiased Multiplex PCR System to Enrich the TRB Repertoire Toward Accurate Detection in Leukemia. Front Immunol 2020; 11:1631. [PMID: 32849555 PMCID: PMC7423970 DOI: 10.3389/fimmu.2020.01631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/18/2020] [Indexed: 11/13/2022] Open
Abstract
Accurate T cell receptor repertoire profiling has provided novel biological and clinical insights in widespread immunological settings; however, there is a lack of reference materials in the community that can be used to calibrate and optimize the various experimental systems in different laboratories. In this study, we designed and synthesized 611 T cell receptor (TCR) beta chain (TRB) templates and used them as reference materials to optimize the multiplex PCR experimental system to enrich the TRB repertoire. We assessed the stability of the optimized system by repeating the experiments in different batches and by remixing the TRB templates in different ratios. These TRB reference materials could be used as independent positive controls to assess the accuracy of the experimental system, and they can also be used as spike-in materials to calibrate the residual biases of the experimental system. We then used the optimized system to detect the minimal residual disease of T cell acute lymphoblastic leukemia and showed a higher sensitivity compared with flow cytometry. We also interrogated how chemotherapy affected the TCR repertoire of patients with B-cell acute lymphoblastic leukemia. Our result shows that high-avidity T cells, such as those targeting known pathogens, are largely selected during chemotherapy, despite the global immunosuppression. These T cells were stimulated and emerged at the time of induction treatment and further expanded during consolidation treatment, possibly to fight against infections. These data demonstrate that accurate immune repertoire information can improve our understanding of the adaptive immunity in leukemia and lead to better treatment management of the patients.
Collapse
Affiliation(s)
- Jinghua Wu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China.,BGI-Shenzhen, Shenzhen, China
| | | | | | | | - Sixi Liu
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Wei Zhang
- BGI-Shenzhen, Shenzhen, China.,Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Lihua Luo
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China.,BGI-Shenzhen, Shenzhen, China
| | | | | | - Yi Zhao
- BGI-Shenzhen, Shenzhen, China
| | - Xiaodong Wang
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Huirong Mai
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Xiuli Yuan
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Feiqiu Wen
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Changgang Li
- Hematology and Oncology Department, Shenzhen Children's Hospital, Shenzhen, China
| | - Xiao Liu
- BGI-Shenzhen, Shenzhen, China.,Neoimmune, Shenzhen, China
| |
Collapse
|
14
|
Mikhael NL, Elsorady M. Clinical significance of T cell receptor excision circle (TREC) quantitation after allogenic HSCT. Blood Res 2019; 54:274-281. [PMID: 31915654 PMCID: PMC6942145 DOI: 10.5045/br.2019.54.4.274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/15/2019] [Accepted: 11/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background Hematopoietic stem cell transplantation (HSCT) is a well-established treatment modality for a variety of diseases. Immune reconstitution is an important event that determines outcomes. The immune recovery of T cells relies on peripheral expansion of mature graft cells, followed by differentiation of donor-derived hematopoietic stem cells. The formation of new T cells occurs in the thymus and as a byproduct, T cell receptor excision circles (TRECs) are released. Detection of TRECs by PCR is a reliable method for estimating the amount of newly formed T cells in the circulation and, indirectly, for estimating thymic function. The aim of this study was to determine the role of TREC quantitation in predicting outcomes of human leucocyte antigen (HLA) identical allogenic HSCT. Methods The study was conducted on 100 patients receiving allogenic HSCT from an HLA identical sibling. TREC quantification was done by real time PCR using a standard curve. Results TREC levels were inversely related to age (P=0.005) and were significantly lower in patients with malignant diseases than in those with benign diseases (P=0.038). TREC levels could predict relapse as an outcome but not graft versus host disease (GvHD) and infections. Conclusion Age and nature of disease determine the TREC levels, which are related to relapse.
Collapse
Affiliation(s)
- Neveen Lewis Mikhael
- Clinical Pathology Department, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Manal Elsorady
- Clinical Hematology Department, Head of BMT Unit, Alexandria Faculty of Medicine, Alexandria, Egypt
| |
Collapse
|
15
|
Finney OC, Brakke H, Rawlings-Rhea S, Hicks R, Doolittle D, Lopez M, Futrell B, Orentas RJ, Li D, Gardner R, Jensen MC. CD19 CAR T cell product and disease attributes predict leukemia remission durability. J Clin Invest 2019; 129:2123-2132. [PMID: 30860496 PMCID: PMC6486329 DOI: 10.1172/jci125423] [Citation(s) in RCA: 249] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells can induce remission in highly refractory leukemia and lymphoma subjects, yet the parameters for achieving sustained relapse-free survival are not fully delineated. METHODS We analyzed 43 pediatric and young adult subjects participating in a Phase I trial of defined composition CD19CAR T cells (NCT02028455). CAR T cell phenotype, function and expansion, as well as starting material T cell repertoire, were analyzed in relation to therapeutic outcome (defined as achieving complete remission within 63 days) and duration of leukemia free survival and B cell aplasia. RESULTS These analyses reveal that initial therapeutic failures (n = 5) were associated with attenuated CAR T cell expansion and/or rapid attrition of functional CAR effector cells following adoptive transfer. The CAR T products were similar in phenotype and function when compared to products resulting in sustained remissions. However, the initial apheresed peripheral blood T cells could be distinguished by an increased frequency of LAG-3+/TNF-αlow CD8 T cells and, following adoptive transfer, the rapid expression of exhaustion markers. For the 38 subjects who achieved an initial sustained MRD-neg remission, remission durability correlated with therapeutic products having increased frequencies of TNF-α-secreting CAR CD8+ T cells, and was dependent on a sufficiently high CD19+ antigen load at time of infusion to trigger CAR T cell proliferation. CONCLUSION These parameters have the potential to prospectively identify patients at risk for therapeutic failure and support the development of approaches to boost CAR T cell activation and proliferation in patients with low levels of CD19 antigen. TRIAL REGISTRATION ClinicalTrials.gov NCT02028455. FUNDING Partial funding for this study was provided by Stand Up to Cancer & St. Baldrick's Pediatric Dream Team Translational Research Grant (SU2C-AACR-DT1113), RO1 CA136551-05, Alex Lemonade Stand Phase I/II Infrastructure Grant, Conquer Cancer Foundation Career Development Award, Washington State Life Sciences Discovery Fund, Ben Towne Foundation, William Lawrence & Blanche Hughes Foundation, and Juno Therapeutics, Inc., a Celgene Company.
Collapse
Affiliation(s)
- Olivia C. Finney
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Hannah Brakke
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Stephanie Rawlings-Rhea
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Roxana Hicks
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Danielle Doolittle
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Marisa Lopez
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Ben Futrell
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rimas J. Orentas
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Daniel Li
- Clinical Statistics Group, Juno Therapeutics, Inc., Seattle, Washington, USA
| | - Rebecca Gardner
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA.,Center for Clinical and Translational Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Michael C. Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
16
|
Das RK, Vernau L, Grupp SA, Barrett DM. Naïve T-cell Deficits at Diagnosis and after Chemotherapy Impair Cell Therapy Potential in Pediatric Cancers. Cancer Discov 2019; 9:492-499. [PMID: 30630850 PMCID: PMC6676489 DOI: 10.1158/2159-8290.cd-18-1314] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/27/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022]
Abstract
Translational data on chimeric antigen receptor (CAR) T-cell trials indicate that the presence of naïve T cells in the premanufacture product is important to clinical response and persistence. In anticipation of developing CAR trials for other tumors, we investigated the T-cell distribution from children with solid tumors and lymphomas at diagnosis and after every cycle of chemotherapy. We found that patients with T cells enriched for naïve and stem central memory cells expanded well in vitro, but the majority of tumor types showed chemotherapy-related depletion of early lineage cells with a corresponding decline in successful ex vivo stimulation response. Unexpectedly, many pediatric patients with solid tumors had low numbers of naïve T cells prior to any therapy. These data indicate the ex vivo manufacture of CAR T cells may need to be customized based on the nature of T cells available in each disease type. SIGNIFICANCE: Cumulative chemotherapy cycles deplete naïve T cells in many pediatric cancer regimens, reducing expansion potential associated with successful adoptive cellular therapies. Naïve T-cell deficits can be seen at diagnosis as well, implying immune deficits that exist prior to chemotherapy, which may also affect the development of immune-based therapies.See related commentary by Leick and Maus, p. 466.This article is highlighted in the In This Issue feature, p. 453.
Collapse
Affiliation(s)
- Rajat K Das
- Division of Oncology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Lauren Vernau
- Division of Oncology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - David M Barrett
- Division of Oncology, Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
17
|
da Rocha LKA, Freschi de Barros S, Bandeira F, Bollini A, Testa LHDA, Simione AJ, Souza MDOE, Zanetti LP, de Oliveira LCS, Dos Santos ACF, de Souza MP, Colturado VAR, Kalil J, Machado CM, Guilherme L. Thymopoiesis in Pre- and Post-Hematopoietic Stem Cell Transplantation. Front Immunol 2018; 9:1889. [PMID: 30245685 PMCID: PMC6137257 DOI: 10.3389/fimmu.2018.01889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is an important therapeutic option for some hematological diseases. However, patients who undergo HSCT acquire a state of immunodeficiency that causes significant mortality. Reconstitution of thymic function is needed to support the immune system. One way to measure thymic function is through T-cell receptor excision circle (TREC) quantification. TRECs are generated by T-cell receptor gene rearrangements during T-cell maturation in the thymus and represent a reliable marker for thymic output. In this study, we aimed to assess aging and malignant hematological diseases as two important factors that may influence thymic output before HSCT. We observed that patients before HSCT presented signal joint TREC (sjTREC) numbers lower than 606.55 copies/μg DNA (low values) compared with healthy individuals, with an odds ratio (OR) of 12.88 [95% confidence interval (CI): 5.26–31.53; p < 0.001]. Our results showed that a group of older individuals (≥50 years old), comprising both healthy individuals and patients, had an OR of 10.07 (95% CI: 2.80–36.20) for low sjTREC values compared with younger individuals (≤24 years old; p < 0.001). Multiple logistic regression analysis confirmed that both older age (≥50 years old) and malignant hematological diseases and their treatments were important and independent risk factors related to thymic function impairment (p < 0.001). The median sjTREC value for patients of all ages was significantly lower than the sjTREC median for the subgroup of older healthy individuals (≥50 years old; p < 0.001). These data suggested that patients before HSCT and healthy individuals exhibited age-dependent thymic impairment, and that prior treatment for hematological diseases may exacerbate aging-related deterioration of natural thymic function. Furthermore, we analyzed these patients 9 months post-HSCT and compared patients who underwent autologous HSCT with those who underwent allogeneic HSCT. Both groups of patients achieved sjTREC copy numbers similar to those of healthy individuals. We did not find a close relationship between impaired thymic function prior to HSCT and worse thymic recovery after HSCT.
Collapse
Affiliation(s)
- Luis Klaus A da Rocha
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Samar Freschi de Barros
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Francine Bandeira
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Alexia Bollini
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | - Anderson João Simione
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Marina de O E Souza
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | - Lilian P Zanetti
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | | | - Mair Pedro de Souza
- Hematopoietic Stem Cell Transplantation Sector, Amaral Carvalho Hospital, Jaú, Brazil
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| | - Clarisse M Machado
- Laboratory of Virology, Tropical Medicine Institute (IMT), University of São Paulo, São Paulo, Brazil
| | - Luiza Guilherme
- Laboratory of Immunology, Heart Institute (InCor), Clinical Hospital, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Ghassemi S, Nunez-Cruz S, O'Connor RS, Fraietta JA, Patel PR, Scholler J, Barrett DM, Lundh SM, Davis MM, Bedoya F, Zhang C, Leferovich J, Lacey SF, Levine BL, Grupp SA, June CH, Melenhorst JJ, Milone MC. Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells. Cancer Immunol Res 2018; 6:1100-1109. [PMID: 30030295 PMCID: PMC8274631 DOI: 10.1158/2326-6066.cir-17-0405] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/22/2017] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
The success of chimeric antigen receptor (CAR)-mediated immunotherapy in acute lymphoblastic leukemia (ALL) highlights the potential of T-cell therapies with directed cytotoxicity against specific tumor antigens. The efficacy of CAR T-cell therapy depends on the engraftment and persistence of T cells following adoptive transfer. Most protocols for T-cell engineering routinely expand T cells ex vivo for 9 to 14 days. Because the potential for engraftment and persistence is related to the state of T-cell differentiation, we hypothesized that reducing the duration of ex vivo culture would limit differentiation and enhance the efficacy of CAR T-cell therapy. We demonstrated that T cells with a CAR-targeting CD19 (CART19) exhibited less differentiation and enhanced effector function in vitro when harvested from cultures at earlier (day 3 or 5) compared with later (day 9) timepoints. We then compared the therapeutic potential of early versus late harvested CART19 in a murine xenograft model of ALL and showed that the antileukemic activity inversely correlated with ex vivo culture time: day 3 harvested cells showed robust tumor control despite using a 6-fold lower dose of CART19, whereas day 9 cells failed to control leukemia at limited cell doses. We also demonstrated the feasibility of an abbreviated culture in a large-scale current good manufacturing practice-compliant process. Limiting the interval between T-cell isolation and CAR treatment is critical for patients with rapidly progressing disease. Generating CAR T cells in less time also improves potency, which is central to the effectiveness of these therapies. Cancer Immunol Res; 6(9); 1100-9. ©2018 AACR.
Collapse
Affiliation(s)
- Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Prachi R Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Scholler
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David M Barrett
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Stefan M Lundh
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Megan M Davis
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Felipe Bedoya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Changfeng Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Leferovich
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Simon F Lacey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci Transl Med 2016; 8:320ra3. [PMID: 26738796 DOI: 10.1126/scitranslmed.aad5222] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Engineered T cell therapies have begun to demonstrate impressive clinical responses in patients with B cell malignancies. Despite this efficacy, many patients are unable to receive T cell therapy because of failure of in vitro expansion, a necessary component of cell manufacture and a predictor of in vivo activity. To evaluate the biology underlying these functional differences, we investigated T cell expansion potential and memory phenotype during chemotherapy in pediatric patients with acute lymphoblastic leukemia (ALL) and non-Hodgkin lymphoma (NHL). We found that patients with T cell populations enriched for early lineage cells expanded better in vitro and that patients with ALL had higher numbers of these cells with a corresponding enhancement in expansion as compared to cells from patients with NHL. We further demonstrated that early lineage cells were selectively depleted by cyclophosphamide and cytarabine chemotherapy and that culture with interleukin-7 (IL-7) and IL-15 enriched select early lineage cells and rescued T cell expansion capability. Thus, early lineage cells are essential to T cell fitness for expansion, and enrichment of this population either by timing of T cell collection or culture method can increase the number of patients eligible to receive highly active engineered cellular therapies.
Collapse
Affiliation(s)
- Nathan Singh
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jessica Perazzelli
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David M Barrett
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Koskenvuo M, Ekman I, Saha E, Salokannel E, Matomäki J, Ilonen J, Kainulainen L, Arola M, Lähteenmäki PM. Immunological Reconstitution in Children After Completing Conventional Chemotherapy of Acute Lymphoblastic Leukemia is Marked by Impaired B-cell Compartment. Pediatr Blood Cancer 2016; 63:1653-6. [PMID: 27163649 DOI: 10.1002/pbc.26047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/04/2016] [Indexed: 11/09/2022]
Abstract
Humoral and cellular immunity were studied in 28 children completing conventional treatment of standard-risk (SR) or intermediate-risk (IR) acute lymphoblastic leukemia (ALL). Both naïve and memory B cells were most severely affected and showed slow recovery during the 2-year follow-up, while the T-cell compartment showed only minor changes. Immunoglobulins and IgG subclasses, components, and antibodies against vaccine-preventable diseases were not significantly affected. In conclusion, immune recovery after conventional chemotherapy for SR and IR ALL is marked by B-cell depletion, but otherwise did not show any severe deficiencies in lymphocyte function.
Collapse
Affiliation(s)
- Minna Koskenvuo
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland.,Division of Pediatric Hematology and Oncology and Stem Cell Transplantation, Department of Pediatrics, Children's Hospital, University of Helsinki, Helsinki, Finland
| | - Ilse Ekman
- Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Emmi Saha
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland
| | - Ellinoora Salokannel
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland
| | - Jaakko Matomäki
- Clinical Research Centre, Turku University Hospital, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, Turku, Finland
| | - Leena Kainulainen
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland
| | - Mikko Arola
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Päivi Maria Lähteenmäki
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Turku University Hospital, University of Turku, Turku, Finland
| |
Collapse
|
21
|
Hanoteau A, Moser M. Chemotherapy and immunotherapy: A close interplay to fight cancer? Oncoimmunology 2016; 5:e1190061. [PMID: 27622046 DOI: 10.1080/2162402x.2016.1190061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/10/2016] [Indexed: 10/21/2022] Open
Abstract
In theory, the immunotherapy of cancer should induce the selective destruction of cancer cells and a long-term specific protection, based on the specificity and memory of immunity. This contrasts with the collateral damages of conventional therapies and their toxic effects on host tissues. However, recent data suggest that chemotherapy may potentiate ongoing immune responses, through homeostatic mechanisms. Massive tumor death, empty "immune" niches and selected cytokines may act as a danger signal, alerting the immune system and amplifying pre-existing antitumor reactivity.
Collapse
Affiliation(s)
- Aurélie Hanoteau
- Laboratory of Immunobiology, Department of Molecular Biology, Université Libre de Bruxelles , Brussels, Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Department of Molecular Biology, Université Libre de Bruxelles , Brussels, Belgium
| |
Collapse
|
22
|
Ingelman-Sundberg HM, Saghafian-Hedengren S, Jahnmatz M, Eksborg S, Jonker M, Nilsson A. Selective loss of vaccine-specific memory B cells in a rhesus macaque model of chemotherapy: influence of doxorubicin on immunological memory. Haematologica 2014; 100:e158-61. [PMID: 25552706 DOI: 10.3324/haematol.2014.116111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | | | | | - Staffan Eksborg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Margreet Jonker
- Biomedical Primate Research Center, Rijswijk, The Netherlands
| | - Anna Nilsson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Öhrmalm L, Smedman C, Wong M, Broliden K, Tolfvenstam T, Norbeck O. Decreased functional T lymphocyte-mediated cytokine responses in patients with chemotherapy-induced neutropenia. J Intern Med 2013; 274:363-70. [PMID: 23789642 DOI: 10.1111/joim.12100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVES The degree of immunosuppression in patients with haematological malignancies treated with chemotherapy is routinely measured as number of circulating cells (preferable neutrophils) in peripheral blood. A parallel decline in the number of T cells is expected, but a possible alteration in their functionality has been less well explored. The ability of T cells to secrete more than one cytokine simultaneously is known to indicate protective immunity. The aim of this study was to determine whether the function of circulating T cells is altered in patients with chemotherapy-induced neutropenia. DESIGN, SETTING AND SUBJECTS In this cross-sectional study, we used the FluoroSpot assay to investigate the proportion of T cells secreting either interferon-γ or interleukin-2, or both cytokines simultaneously, after anti-CD3 stimulation. Peripheral blood mononuclear cells from 53 adult patients with chemotherapy-induced neutropenia and 20 healthy individuals were investigated. RESULTS There were significantly fewer T cells secreting interferon-γ in patients with neutropenia compared with healthy control subjects (P = 0.02), but the difference was greatest for dual cytokine-secreting T cells (P = 0.001). Furthermore, the amount of secreted cytokine per T cell appeared to be reduced in patients, compared with control subjects. CONCLUSION Our results suggest that the functionality of T cells is altered in patients with haematological malignancies with chemotherapy-induced neutropenia. In parallel with a decline in T cell count, this may further increase the risk of severe infections.
Collapse
Affiliation(s)
- L Öhrmalm
- Department of Medicine, Solna, Infectious Diseases Unit, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
24
|
Halac U, Béland K, Lapierre P, Patey N, Ward P, Brassard J, Houde A, Alvarez F. Cirrhosis due to chronic hepatitis E infection in a child post-bone marrow transplant. J Pediatr 2012; 160:871-4.e1. [PMID: 22341950 DOI: 10.1016/j.jpeds.2012.01.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 10/25/2011] [Accepted: 01/12/2012] [Indexed: 01/01/2023]
Abstract
Chronic hepatitis E virus (HEV) infection occurs in immunosuppressed adults. We detected HEV ribonucleic acid in serum of an adolescent patient who had undergone bone marrow transplantation and subsequently presented with persistently increased aminotransferases and histologic chronic hepatitis, and eventually developed cirrhosis. Phylogenetic analysis revealed these HEV strains were similar to swine genotype 3a, suggesting a possible zoonosis.
Collapse
Affiliation(s)
- Ugur Halac
- Department of Pediatrics, Université de Montréal, Quebec, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Vaccination in children with cancer: a debate. Open Med (Wars) 2012. [DOI: 10.2478/s11536-011-0143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AbstractChildren with malignancy present an important alteration of the immune system functionality caused by the illness itself and by the therapy they undergo. Therefore, they are at high risk of contracting vaccine-preventable diseases and of developing important complications. Vaccinations represent valid devices against these infections but this condition involves two main problems: are vaccines safe in these patients? Are vaccines effective in these patients? The aim of this review is to discuss the principles of vaccination management in children with cancer.
Collapse
|
26
|
Shetty AK, Winter MA. Immunization of children receiving immunosuppressive therapy for cancer or hematopoietic stem cell transplantation. Ochsner J 2012; 12:228-243. [PMID: 23049460 PMCID: PMC3448245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
In the past 3 decades, the number of immunocompromised children has increased steadily because of dramatic improvement in survival rates in certain malignancies as a result of intensive curative treatment regimens and an increase in the number of children undergoing life-saving hematopoietic stem cell transplantation (HSCT). Children receiving immunosuppressive therapy for cancer, as well as HSCT recipients, will benefit from vaccination but warrant close evaluation for a variety of reasons, such as the risk of developing severe infections, serious adverse events following certain vaccines, and decreased vaccine efficacy caused by poor immune response to vaccination. Various professional organizations have published vaccination guidelines for immunocompromised patients. Given their heterogeneity, recommendations for the immunization of immunocompromised patients may not be universally applicable. The safety of many commonly used vaccines has not been established in immunocompromised children. In addition, no large-scale vaccine studies have evaluated the clinical outcome of disease prevention in this population. All killed vaccines are generally safe, while live vaccines may be administered to immunocompromised children in select circumstances, depending on the degree of altered immunocompetence and the underlying primary condition. Healthcare providers should be knowledgeable about the indications, contraindications, and precautions for vaccine administration in immunocompromised patients. To protect immunocompromised patients, all family, household contacts, and healthcare workers should also be immunized with all routinely recommended vaccines. Pediatricians play a crucial role in identifying and effectively communicating the risks and benefits of vaccines to immunocompromised patients and their parents.
Collapse
Affiliation(s)
| | - Mary A. Winter
- Department of Pharmacy Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
27
|
Ruggiero A, Battista A, Coccia P, Attinà G, Riccardi R. How to manage vaccinations in children with cancer. Pediatr Blood Cancer 2011; 57:1104-8. [PMID: 21953691 DOI: 10.1002/pbc.23333] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 08/08/2011] [Indexed: 11/07/2022]
Abstract
The optimal use of routine childhood immunizations in children with malignancy is still a matter of debate. Despite their higher risk of contracting vaccine preventable diseases and of suffering important complications, there is little understanding of the magnitude of the possible benefit of administering active immunization in this population due to a paucity of clinical trial data. Our review focuses on the management of children with cancer and offers some suggestions regarding their vaccination schedules.
Collapse
Affiliation(s)
- Antonio Ruggiero
- Pediatric Oncology Division, A Gemelli Hospital, Catholic University, Rome, Italy.
| | | | | | | | | |
Collapse
|
28
|
Ek T, Josefson M, Abrahamsson J. Multivariate analysis of the relation between immune dysfunction and treatment intensity in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2011; 56:1078-87. [PMID: 21344616 DOI: 10.1002/pbc.23043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 12/27/2010] [Indexed: 11/09/2022]
Abstract
BACKGROUND Immunoreconstitution following childhood acute lymphoblastic leukemia (ALL) is a complex process during which various immune functions recover differentially. This process is difficult to elucidate since variables are interrelated and require simultaneous evaluation, rendering conventional statistical methods inappropriate. PROCEDURE We used principal components analysis (PCA) and projection of latent structures (PLS) to evaluate immune competence in 32 children treated for ALL. One or 6 months after completion of therapy, the relation between lymphocyte subpopulations, lymphocyte function and response to vaccination with tetanus, diphtheria and hemophilus influenzae, was investigated. RESULTS PCA demonstrated that increasing treatment intensity correlated with progressive immune dysfunction. Children treated with high intensity had poor response to vaccination associated with loss of humoral memory, decreased CD4(+) 45RA(+) T-lymphocytes and increased CD5+ B-lymphocytes. Patients treated with intermediate intensity had better preservation of humoral memory but decreased CD4(+) 45RA(+) T-cells. Patients with a low intensity regimen had similar vaccination response and lymphocyte levels as controls. CONCLUSIONS Our findings demonstrate the utility of PCA and PLS in detecting hidden structures in complex data and suggest that, even 6 months after therapy, patients treated with intermediate and high intensity have attenuated responses to de novo antigens whereas those with high intensity also respond poorly to recall antigens.
Collapse
Affiliation(s)
- Torben Ek
- Department of Paediatrics, Institution for Clinical Sciences, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | |
Collapse
|
29
|
van Tilburg CM, van der Velden VH, Sanders EA, Wolfs TF, Gaiser JF, de Haas V, Pieters R, Bloem AC, Bierings MB. Reduced versus intensive chemotherapy for childhood acute lymphoblastic leukemia: Impact on lymphocyte compartment composition. Leuk Res 2011; 35:484-91. [DOI: 10.1016/j.leukres.2010.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 10/10/2010] [Accepted: 10/11/2010] [Indexed: 01/18/2023]
|
30
|
van Tilburg CM, Sanders EA, Nibbelke EE, Pieters R, Revesz T, Westers P, Wolfs TF, Bierings MB. Impact of reduced chemotherapy treatment for good risk childhood acute lymphoblastic leukaemia on infectious morbidity*. Br J Haematol 2011; 152:433-40. [DOI: 10.1111/j.1365-2141.2010.08463.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
31
|
Leung W, Neale G, Behm F, Iyengar R, Finkelstein D, Kastan MB, Pui CH. Deficient innate immunity, thymopoiesis, and gene expression response to radiation in survivors of childhood acute lymphoblastic leukemia. Cancer Epidemiol 2010; 34:303-8. [PMID: 20413363 DOI: 10.1016/j.canep.2010.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Accepted: 03/15/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Survivors of childhood acute lymphoblastic leukemia (ALL) are at an increased risk of developing secondary malignant neoplasms. Radiation and chemotherapy can cause mutations and cytogenetic abnormalities and induce genomic instability. Host immunity and appropriate DNA damage responses are critical inhibitors of carcinogenesis. Therefore, we sought to determine the long-term effects of ALL treatment on immune function and response to DNA damage. METHODS Comparative studies on 14 survivors in first complete remission and 16 siblings were conducted. RESULTS In comparison to siblings on the cells that were involved in adaptive immunity, the patients had either higher numbers (CD19+ B cells and CD4+CD25+ T regulatory cells) or similar numbers (alphabetaT cells and CD45RO+/RA- memory T cells) in the blood. In contrast, patients had lower numbers of all lymphocyte subsets involved in innate immunity (gammadeltaT cells and all NK subsets, including KIR2DL1+ cells, KIR2DL2/L3+ cells, and CD16+ cells), and lower natural cytotoxicity against K562 leukemia cells. Thymopoiesis was lower in patients, as demonstrated by less CD45RO-/RA+ naïve T cell and less SjTREC levels in the blood, whereas the Vbeta spectratype complexity score was similar. Array of gene expression response to low-dose radiation showed that about 70% of the probesets had a reduced response in patients. One of these genes, SCHIP-1, was also among the top-ranked single nucleotide polymorphisms (SNPs) during the whole-genome scanning by SNP microarray analysis. CONCLUSION ALL survivors were deficient in innate immunity, thymopoiesis, and DNA damage responses to radiation. These defects may contribute to their increased likelihood of second malignancy.
Collapse
Affiliation(s)
- Wing Leung
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-2794, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Cheng FWT, Leung TF, Chan PKS, Leung WK, Lee V, Shing MK, Yuen PMP, Li CK. Recovery of humoral and cellular immunities to vaccine-preventable infectious diseases in pediatric oncology patients. Pediatr Hematol Oncol 2010; 27:195-204. [PMID: 20367263 DOI: 10.3109/08880011003621752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The recovery of antibodies to various vaccine-preventable infectious diseases, humoral and cellular immunity in pediatric oncology patients were evaluated by a prospective longitudinal study for 18 months. Lymphocyte subset (CD3+, CD4+, CD8+, CD16/56+, CD19+), CD4/CD8 ratio, immunoglobulin levels, antibodies to diphtheria, pertussis, tetanus, hepatitis B, measles, mumps, and rubella were measured serially at 6 months till 18 months after stopping all chemotherapy (including maintenance chemotherapy). Twenty-eight children (hematological malignancies, n = 14; solid tumors, n = 14) were studied. The median age was 7.0 +/- 3.8 years old (range 2.6-16.2 years old). Although there was significant increase in CD3+, CD4+, CD8+, CD19+ cells, IgG, IgA, and IgM levels (P < .05), CD4+ and CD8+ counts were still below the age-specific normal range at the end of study period. At 18 months after stopping chemotherapy, 11%, 15%, 60%, 30%, 49%, and 30% of subjects remained seronegative against diphtheria, tetanus, hepatitis B, measles, mumps, and rubella. This will evolve to a significant health care problem if no further intervention is implemented, as the survival rate of pediatric oncology patients improves significantly with the improvement in various cancer treatment protocols. Near complete immune recovery was demonstrated in the subjects. Significant proportion of subjects remained susceptible to vaccine-preventable infectious diseases up to 18 months after stopping all chemotherapy.
Collapse
Affiliation(s)
- Frankie Wai Tsoi Cheng
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Eyrich M, Wiegering V, Lim A, Schrauder A, Winkler B, Schlegel PG. Immune function in children under chemotherapy for standard risk acute lymphoblastic leukaemia - a prospective study of 20 paediatric patients. Br J Haematol 2009; 147:360-70. [DOI: 10.1111/j.1365-2141.2009.07862.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Fioredda F, Cavillo M, Banov L, Plebani A, Timitilli A, Castagnola E. Immunization after the elective end of antineoplastic chemotherapy in children. Pediatr Blood Cancer 2009; 52:165-8. [PMID: 19034908 DOI: 10.1002/pbc.21864] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The aim of the present commentary is to discuss the multifaceted topic of vaccinations after treatment for cancer in the pediatric age. Publications in this field reveal conflicting data and opinions; no evidence-based guidelines currently exist. However, in spite of several discrepancies some commonly accepted information and conclusions exist. Efforts to find a common strategy of re-immunization should be directed towards setting up prospective studies on sufficient numbers of patients to obtain statistically relevant end points.
Collapse
Affiliation(s)
- Francesca Fioredda
- Haematology Unit, Department of Haematology and Oncology, "G.Gaslini" Children Hospital, Genoa, Italy.
| | | | | | | | | | | |
Collapse
|
35
|
Greiner J, Bullinger L, Guinn BA, Döhner H, Schmitt M. Leukemia-associated antigens are critical for the proliferation of acute myeloid leukemia cells. Clin Cancer Res 2008; 14:7161-6. [PMID: 19010831 DOI: 10.1158/1078-0432.ccr-08-1102] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. With intensive induction therapy, most patients younger than 60 years achieve complete remission. However, even if these younger patients were treated intensively, more than 50% will relapse. Clinical results of patients older than 60 years are more unfavorable. Therefore, in all patients with AML, the overall survival is still low. In the past decade, several leukemia-associated antigens (LAA) have been identified in patients with acute myeloid leukemia. BAGE, BCL-2, OFA-iLRP, FLT3-ITD, G250, hTERT, PRAME, proteinase 3, RHAMM, survivin, and WT-1 are all LAAs that have been shown to induce CD8+ T-cell recognition and for some antigens also humoral immune responses. Interestingly, most of these LAAs are linked to cell cycle or proliferation. This article discusses the balance between LAA-driven leukemia cell expansion and the elimination of these cells through attacks on LAAs by the immune system. Current knowledge of the function and CD8+ T-cell recognition of LAAs is reviewed and an outlook is given on how to improve T-cell responses to LAAs in acute myeloid leukemia cells.
Collapse
Affiliation(s)
- Jochen Greiner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany.
| | | | | | | | | |
Collapse
|
36
|
Lehrnbecher T, Koehl U, Wittekindt B, Bochennek K, Tramsen L, Klingebiel T, Chanock SJ. Changes in host defence induced by malignancies and antineoplastic treatment: implication for immunotherapeutic strategies. Lancet Oncol 2008; 9:269-78. [DOI: 10.1016/s1470-2045(08)70071-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
37
|
Chaput N, De Botton S, Obeid M, Apetoh L, Ghiringhelli F, Panaretakis T, Flament C, Zitvogel L, Kroemer G. Molecular determinants of immunogenic cell death: surface exposure of calreticulin makes the difference. J Mol Med (Berl) 2007; 85:1069-76. [PMID: 17891368 DOI: 10.1007/s00109-007-0214-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 04/23/2007] [Accepted: 04/25/2007] [Indexed: 12/17/2022]
Abstract
The treatment of cancer by chemotherapy causes tumour cell death, mostly by apoptosis. This tumour cell death may or may not elicit an immune response. At least in some cases, the efficacy of chemotherapy critically depends on the induction of immunogenic cell death that is a type of cell demise that stimulates the activation of an adaptative anti-tumour immune response, which in turn helps to eradicate residual cancer (stem) cells. Indeed, anthracyclins care more efficient in curing tumours in immunocompetent than in T cell-deficient mice. The molecular mechanism implicated in this anti-tumour T cell activation was recently discovered. Anthracyclins cause immunogenic cell death due to their specific capacity to stimulate the translocation of calreticulin to the cell surface. Calreticulin then acts as an "eat me" signal for dendritic cells, allowing them to phagocytose tumour cells and to prime tumour antigen-specific cytotoxic T cells. Importantly, non-immunogenic chemotherapy can be rendered immunogenic by adsorbing recombinant calreticulin to tumour cells or by enforcing the translocation of endogenous calreticulin to the cell surface by means of PP1/GADD34 inhibitors. This strategy could have major implications for the treatment of human cancer. Indeed, in vivo treatments with anthracyclins can cause the translocation of calreticulin to the surface of circulating tumour cells, in patients with acute myeloid leukaemia (AML). The challenge will be to determine whether the exposure of calreticulin translocation on the tumour cell surface is linked to chemotherapy-induced anti-tumour immune responses and therapeutic efficacy in human cancer.
Collapse
MESH Headings
- Animals
- Anthracyclines/pharmacology
- Anthracyclines/therapeutic use
- Antigens, Neoplasm/drug effects
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Surface/drug effects
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/immunology
- Calreticulin/immunology
- Calreticulin/metabolism
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Dendritic Cells/immunology
- Humans
- Mice
- Models, Biological
- Neoplasms, Experimental/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Nathalie Chaput
- Centre d'Investigation Clinique Biothérapie, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Coughlin CM, Fleming MD, Carroll RG, Pawel BR, Hogarty MD, Shan X, Vance BA, Cohen JN, Jairaj S, Lord EM, Wexler MH, Danet-Desnoyers GAH, Pinkus JL, Pinkus GS, Maris JM, Grupp SA, Vonderheide RH. Immunosurveillance and Survivin-Specific T-Cell Immunity in Children With High-Risk Neuroblastoma. J Clin Oncol 2006; 24:5725-34. [PMID: 17179106 DOI: 10.1200/jco.2005.05.3314] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PurposeTumor immunosurveillance influences oncogenesis and tumor growth, but it remains controversial whether clinical failure of immunosurveillance is a result of lymphocyte dysfunction or tumor escape. In this study, our goal was to characterize the physiology of tumor immunosurveillance in children with high-risk neuroblastoma (HR-NBL).Patients and MethodsImmunohistopathologic studies were carried out on 26 tumor samples from a cohort of HR-NBL patients diagnosed at Children's Hospital of Philadelphia for the 2-year period from May 2003 to May 2005. Blood from nine HLA-A2+patients in this cohort was analyzed for T cells specific for the antiapoptotic protein survivin.ResultsSurvivin protein was expressed by 26 of 26 tumors. In HLA-A2+patients, circulating cytotoxic T lymphocytes (CTLs) specific for survivin were detected by peptide/major histocompatibility complex tetramer analysis in the blood of eight of nine children with HR-NBL at the time of diagnosis. Rather than being selectively rendered anergic in vivo, circulating survivin-specific CTLs were highly functional as shown by cytotoxicity and interferon gamma enzyme-linked immunospot assays in six of nine patients. Survivin-specific CD107a mobilization by T cells was found in five of five patients. By immunohistochemistry, tumor-infiltrating T cells were few or absent in 26 of 26 tumors.ConclusionChildren with HR-NBL harbor robust cellular immune responses to the universal tumor antigen survivin at the time of diagnosis, but intratumoral T cells are strikingly rare, suggesting a failure of cellular immunosurveillance. Efforts to develop novel therapies that increase T-cell trafficking into tumor nests are warranted.
Collapse
Affiliation(s)
- Christina M Coughlin
- Abramson Family Cancer Research Institute, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yetgin S, Tavil B, Aytac S, Kuskonmaz B, Kanra G, Karna G. Unexpected protection from infection by two booster hepatitis B virus vaccination in children with acute lymphoblastic leukemia. Leuk Res 2006; 31:493-6. [PMID: 16930691 DOI: 10.1016/j.leukres.2006.06.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Revised: 06/16/2006] [Accepted: 06/16/2006] [Indexed: 11/17/2022]
Abstract
The protective power of two booster dose vaccination against hepatitis B virus (HBV) infection has not been previously studied in patients with acute lymphoblastic leukemia (ALL) who remained unresponsive to immunization. The aim of this study was to determine the HBV infection rate in vaccinated and unvaccinated patients with or without seroconversion and to compare these groups in respect to HBV infection rate. The study group included 111 male and 85 female ALL patients with a mean age of 6.23+/-4.10 years. Patients were divided into three groups as follows: Group 1 included 82 patients who were vaccinated during maintenance chemotherapy, Group 2 included 87 unvaccinated patients, and Group 3 included 27 patients who were vaccinated prior to the diagnosis of ALL. Seroconversion was obtained in 35.4% (29/82) of patients in Group 1. The incidence of HBV infection was significantly lower in Group 1 (4/82, 4.8%) than in Group 2 (25/87, 28.7%). When we compared only the seronegative patients in Group 1 with Group 2 in respect to HBV infection rate, Group 1 still had a significantly lower HBV infection rate than Group 2 (7.5% versus 28.7%) (p<0.001). No patients in Group 3 (n=27) had HBV infection. In addition to the seroconversion level, infection rate is also important in the evaluation of the effectiveness of vaccination. Our study results suggest that a high protective role of HBV vaccination was also observed in non-seroconversion ALL patients. The effect of cellular immunity on the protection against infection should also be investigated in such patients with further studies.
Collapse
Affiliation(s)
- Sevgi Yetgin
- Department of Pediatric Hematology, Hacettepe University Faculty of Medicine, Sihhiye, Ankara 06100, Turkey.
| | | | | | | | | | | |
Collapse
|
40
|
van Tilburg CM, Sanders EAM, Rovers MM, Wolfs TFW, Bierings MB. Loss of antibodies and response to (re-)vaccination in children after treatment for acute lymphocytic leukemia: a systematic review. Leukemia 2006; 20:1717-22. [PMID: 16888619 DOI: 10.1038/sj.leu.2404326] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intensified chemotherapy regimens resulting in improved survival of children with acute lymphocytic leukemia (ALL) lead to concerns about therapy-induced immune damage reflected by the loss of protection of previous immunizations and the efficacy of (re-)vaccination. The severity of secondary immunodeficiency, however, is not clear and knowledge is based on a limited number of studies. We performed a systematic review on literature concerning vaccination data of children with ALL published since 1980. Eight studies fulfilled the inclusion criteria. Regarding antibody titers after treatment, the number of children who had preserved the defined protection level for antibodies differed widely, ranging from 17 to 98% for diphtheria, 27 to 82% for Bordetella pertussis, 20 to 98% for tetanus, 62 to 100% for poliomyelitis, 35 to 100% for Haemophilus influenzae type B (HiB), 29 to 92% for mumps, 29 to 60% for measles and 72 to 92% for rubella. Most patients however responded to revaccination, demonstrating immunological recovery. Although the designs and results of the included studies varied widely, it can be concluded that cytostatic therapy for ALL in children results in a temporarily reduction of specific antibody levels. Memory is preserved but revaccination may be warranted. This is the first systematic review and the best possible current approximation of chemotherapy-induced immune damage in children after ALL treatment.
Collapse
Affiliation(s)
- C M van Tilburg
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
41
|
Current Awareness in Hematological Oncology. Hematol Oncol 2006. [DOI: 10.1002/hon.752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
42
|
Rousseau RF, Biagi E, Dutour A, Yvon ES, Brown MP, Lin T, Mei Z, Grilley B, Popek E, Heslop HE, Gee AP, Krance RA, Popat U, Carrum G, Margolin JF, Brenner MK. Immunotherapy of high-risk acute leukemia with a recipient (autologous) vaccine expressing transgenic human CD40L and IL-2 after chemotherapy and allogeneic stem cell transplantation. Blood 2005; 107:1332-41. [PMID: 16249392 PMCID: PMC1895421 DOI: 10.1182/blood-2005-03-1259] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD40L generates immune responses in leukemia-bearing mice, an effect that is potentiated by IL-2. We studied the feasibility, safety, and immunologic efficacy of an IL-2- and CD40L-expressing recipient-derived tumor vaccine consisting of leukemic blasts admixed with skin fibroblasts transduced with adenoviral vectors encoding human IL-2 (hIL-2) and hCD40L. Ten patients (including 7 children) with high-risk acute myeloid (n = 4) or lymphoblastic (n = 6) leukemia in cytologic remission (after allogeneic stem cell transplantation [n = 9] or chemotherapy alone [n = 1]) received up to 6 subcutaneous injections of the IL-2/CD40L vaccine. None of the patients were receiving immunosuppressive drugs. No severe adverse reactions were noted. Immunization produced a 10- to 890-fold increase in the frequencies of major histocompatibility complex (MHC)-restricted T cells reactive against recipient-derived blasts. These leukemia-reactive T cells included both T-cytotoxic/T-helper 1 (Th1) and Th2 subclasses, as determined from their production of granzyme B, interferon-gamma, and interleukin-5. Two patients produced systemic IgG antibodies that bound to their blasts. Eight patients remained disease free for 27 to 62 months after treatment (5-year overall survival, 90%). Thus, even in heavily treated patients, including recipients of allogeneic stem cell transplants, recipient-derived antileukemia vaccines can induce immune responses reactive against leukemic blasts. This approach may be worthy of further study, particularly in patients with a high risk of relapse.
Collapse
Affiliation(s)
- Raphaël F Rousseau
- Center for Cell and Gene Therapy, 6621 Fannin St, MC3-3320, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|